Prospects for oral replicating adenovirus

Vaccine 31 (2013) 3236–3243
Contents lists available at SciVerse ScienceDirect
Vaccine
journal homepage: www.elsevier.com/locate/vaccine
Review
Prospects for oral replicating adenovirus-vectored vaccines
Cailin Deal, Andrew Pekosz, Gary Ketner ∗
W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United
States
a b s t r a c t
i n f o
DR
a r t i c l e
Article history:
Received 1 March 2013
Received in revised form 6 May 2013
Accepted 7 May 2013
Available online 22 May 2013
or
C
Orally delivered replicating adenovirus (Ad) vaccines have been used for decades to prevent adenovirus
serotype 4 and 7 respiratory illness in military recruits, demonstrating exemplary safety and high efficacy. That experience suggests that oral administration of live recombinant Ads (rAds) holds promise
for immunization against other infectious diseases, including those that have been refractory to traditional vaccination methods. Live rAds can express intact antigens from free-standing transgenes during
replication in infected cells. Alternatively, antigenic epitopes can be displayed on the rAd capsid itself,
allowing presentation of the epitope to the immune system both prior to and during replication of the
virus. Such capsid-display rAds offer a novel vaccine approach that could be used either independently of
or in combination with transgene expression strategies to provide a new tool in the search for protection
from infectious disease.
© 2013 Elsevier Ltd. All rights reserved.
or
iz
ad
ap
Keywords:
Replicating adenovirus vectors
Oral administration
Capsid-display
Hexon
Vaccine
Contents
1.
2.
3.
Co
pi
aa
ut
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3237
Replicating rAd transgene vectors as vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3237
Replicating capsid display rAds as vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3238
3.1.
Hexon (polypeptide II) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3238
3.2.
Penton base (polypeptide III) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3239
3.3.
Fiber (polypeptide IV) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3239
3.4.
pIX (polypeptide IX) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3240
3.5.
Comparative immunogenicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3240
3.6.
Pre-existing immunity and replicating rAds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3241
4.
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3241
Author contributions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3241
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3241
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3241
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3241
Abbreviations: Ad, adenovirus; rAd, recombinant adenovirus; WT, wild-type; E1, early region 1; E4, early region 4; HBsAg, hepatitis B virus surface antigen; hr404, host
range mutation in DNA binding protein; rAd5hr, recombinant Ad 5 with hr404 mutation; Ad5hrSIVenv/rev, Ad5 with hr404 mutation expressing env and rev genes of SIV;
rAd4-H5-Vtn, recombinant Ad4 expressing influenza H5 hemagglutinin; HA, influenza hemagglutinin; VP, virus particles; MHC class II, major histocompatibility complex
II; MHC class I, major histocompatibility complex I; HVRs, hypervariable regions; nAb, neutralizing antibodies; PEI, preexisting immunity; OprF, outer membrane protein F
of Pseudomonas aeruginosa; PA, protective antigen of Bacillus anthracis; CSP, Plasmodium circumsporozoite protein; M2e, influenza A matrix protein 2 extracellular domain;
VR1, variable region 1; RGD, Arg-Gly-Asp motif; FV, friend murine leukemia virus; OVA, ovalbumin.
∗ Corresponding author. Tel.: +1 410 955 3776; fax: +1 410 955 0105.
E-mail address: [email protected] (G. Ketner).
0264-410X/$ – see front matter © 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.vaccine.2013.05.016
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
Co
pi
aa
ut
or
C
ap
or
iz
ad
Oral delivery of immunogens to the gut is regarded as the “Holy
Grail” for vaccinologists [1]. The intestine is the largest lymphoid
organ and gut-associated immune cells represent up to 90% of
immunocompetent cells [2]. Oral immunization offers immunological and logistical advantages including stimulation of mucosal
immune responses preferentially at the site of entry for many infectious agents and ability to elicit strong systemic immunity. Oral
immunization is cost effective and offers improved patient compliance due to the ease of vaccine administration, freedom from
needles and from the requirement for trained medical personnel.
All three oral vaccines licensed for use in the US [3] contain live
virus. Live-virus vaccines add to the inherent advantages of oral
immunization the ability to immunize with small (and hence less
expensive) doses, and induction of a breadth of immune responses
similar to those induced by natural infection. These characteristics
would facilitate routine immunization and response to epidemics
or pandemics [4] and make live oral immunization attractive in
resource-poor regions, where economy and logistical tractability
are critically important.
Licensed oral adenovirus (Ad) serotype 4 and 7 vaccines provide
a model for use of live recombinant adenoviruses (rAds) for oral
immunization. Since the 1970s, live oral Ad vaccines have been used
by the United States military to prevent acute respiratory disease
caused by Ad4 and Ad7 [5]. These vaccines contain lyophilized live,
wild type (WT) virus incorporated into enteric tablets that protect
the virus against the low pH of the stomach. After oral administration of the tablets, live virus is released into the intestine where
asymptomatic replication occurs. In a single dose, the vaccines generate an immune response that was over 95% effective in preventing
Ad4- and Ad7-induced respiratory illness in a clinical trial involving
more than 40,000 soldiers [6–9]. The historical success of Ad military vaccines suggests great potential for recombinant vaccines
using the oral replicating Ad platform.
rAds have been used to deliver vaccine antigens in over 90 preclinical and clinical trials [10,11]. The rationales for use of rAd
vaccines include genome stability and ease of manipulation, natural tropism for mucosal inductive sites including the gut and upper
respiratory tract and ability to elicit vigorous humoral and cellular
immune responses. rAds infect a broad spectrum of cells, including dendritic cells, allowing for efficient antigen presentation and
can therefore also prime a robust cell-mediated response [12,13].
However, most rAd vaccine candidates are replication defective
and not intended for oral administration. Here, we review work
on replicating rAd vaccines that may provide a route to effective
oral immunization.
support replication of some human Ads, providing systems that
might be exploited to test replicating vaccines [19–24]. Cotton
rats and hamsters have found use in characterization of replicating oncolytic adenoviruses [19,25], and dogs have been used
in evaluation of live rAd vaccines [21]. In practice, however,
well-developed immunological reagents, perceived similarity of
primate and human immune responses, and availability of suitable challenges to assess efficacy have restricted most studies of
replication-competent rAds in permissive hosts to primates (chimpanzees or monkeys), or to human volunteers.
In early studies, replication-competent rAd7 and rAd4 expressing the hepatitis B virus surface antigen (HBsAg) were used
to immunize (rAd7 HBsAg) and then boost (rAd4 HBsAg) two
Ad4, Ad7-seronegative chimpanzees (rAd7/rAd4 HBsAg) by the
oral route [23]. After primary vaccinations, both chimpanzees
shed vaccine virus for 6–7 weeks and developed Ad7 antibodies, suggesting successful Ad7 replication in the chimpanzee
gut. One developed transient seropositivity for HBsAg after the
first inoculation; both developed modest titers after the second.
A third chimpanzee immunized with WT Ad7 and then rAd4
HBsAg (WTAd7/rAd4 HBsAg) developed no HBsAg antibodies. Both
rAd7/rAd4 HBsAg chimpanzees were protected from acute clinical
disease but were not protected from infection as evident by development of antibodies against the HBV core protein in response to
HBV challenge. The animal that did not seroconvert (WTAd7/rAd4
HBsAg), along with an unimmunized control, became clinically
infected with HBV [23]. Three human volunteers in a small phase
I vaccine trial immunized with the rAd7 HBsAg vaccine exhibited no adverse effects and shed virus between days 4 and 13
post vaccination with no evidence of person-to-person spread.
Although all subjects had a significant increase in Ad7 antibodies, none made antibodies to HBsAg [26]. Protection from disease,
if not infection, in chimpanzees, despite lack of seroconversion
in humans, suggests potential value in using oral enteric vaccination with rAd to induce humoral immune responses to foreign
pathogens.
Most animal studies of replicating rAds have been conducted
in macaques. WT Ad2 and Ad5 do not replicate in monkeys, and
these experiments therefore require use of an Ad5 host range mutation (hr404), located in the 72k DNA binding protein, that permits
replication in monkey cells and macaques [24,27]. A transgenetype rAd5 hr404 (rAd5hr) virus expressing the env and rev genes
from SIV (Ad5hr-SIVenv/rev) was able to replicate in vivo in rhesus macaques [28]. Priming orally and intranasally, followed by
intratracheal immunization 12 weeks later with Ad5hr-SIVenv/rev,
generated proliferating T cells to Env and strong serum neutralizing
anti-Env antibodies. Mucosal secretions also contained Env-specific
IgG and IgA antibodies. Although this vaccine did not induce sterilizing immunity, it conferred acute-phase protection following
intravaginal challenge with SIV [28]. Partial protection of reboosted
and rechallenged transiently viremic macaques was associated
with both cellular and humoral immune responses [29]. To broaden
rAd-induced immunity to SIV, additional rhesus macaques were
immunized simultaneously with replicating constructs expressing
SIV env, rev and gag genes through oral and intranasal administration [30]. Specific T-cell responses were generated against all SIV
gene products and there was a persistent response to Gag evident
for more than 10 weeks post-immunization. Interestingly, immunization primed CD8+ T cells for a persistent and potent response
to both dominant and subdominant epitopes [30,31]. Intrarectal
challenge with SIV demonstrated that the vaccine did not induce
sterile immunity but acute viral replication was suppressed. Cellular immunity to SIV Gag and Env, along with nasal and vaginal
Env-specific IgG antibodies, correlated with a significant reduction
of acute phase viremia [32]. Immunized groups exhibited significant protection, with 39% of macaques having either no viremia,
DR
1. Introduction
3237
2. Replicating rAd transgene vectors as vaccines
Most current rAd vaccine candidates are transgene expression
vectors, commonly engineered to express a foreign gene inserted
into early region 1 (E1) or, occasionally, early region 4 (E4) of the
genome [14]. E1 and E4 are essential for viral replication, and most
such rAds are replication-defective [15–17]. Extensive experience
with defective recombinants in humans and animal models has
shown promise in several cases [18].
Replication-competent transgene vectors can be constructed by
careful choice of the site of transgene insertion but relatively few
have been extensively investigated. Study of replicating rAd vaccines is complicated by the requirement for a host that supports
viral replication if vaccines are to be evaluated under conditions
that mimic their intended use in humans. Mice do not support
human adenovirus replication. However, golden hamsters, cotton rats, dogs, pigs, monkeys (see below), and chimpanzees all
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
antigens are incorporated into one of the capsid proteins such that
they are displayed on the surface of the virus particle. Capsidincorporated antigens are available for binding by surface antibody
on B cells and can be processed by the exogenous (MHC class II)
pathway. Thus, capsid display recombinants can be immunogenic
without intracellular antigen expression, including in systems that
do not support virus replication. Replication in a permissive host
would further allow persistent antigen presentation via both the
exogenous and the endogenous (MHC class I) pathways, with the
potential of inducing both humoral and cellular responses. Capsiddisplay vectors are extremely immunogenic in mice [41–43] and
therefore may offer greater efficacy in inducing humoral responses
in permissive systems than do transgene rAds.
Several capsid proteins can display foreign epitopes, including
hexon, fiber, penton base and pIX (Table 1 and Fig. 1A). Currently,
immunogenicity data is available only in mice, and conclusions
therefore have been drawn only in the absence of viral replication.
3.1. Hexon (polypeptide II)
or
C
DR
The ∼960 amino acid Ad hexon protein is the most abundant of
the capsid proteins, present in 720 copies per particle [44]. Analysis
of hexon amino acid sequences from different serotypes revealed 9
hypervariable regions (HVRs) that diverge in sequence and length
among serotypes [45]. Crystal structures of Ad2 and Ad5 hexon
show that HVRs reside in two loops that form the surface-exposed
portion of hexon. HVR 1–6 are located within the DEI loop and
HVR 7–9 lie within the FGI loop (Fig. 1B and D) [45,46]. These
HVRs contain serotype-specific epitopes that are primary targets
of neutralizing antibodies (nAb) [47].
X-ray crystallography suggests that HVRs 2, 3, 5–7 are
unordered and protrude from the capsid surface. Virus containing
insertions of His6 peptides with flanking spacers into those HVRs
are viable, with normal virion thermostability and infectivity [48].
His6 in HVR2 or 5 is capable of binding tightly to the His6 antibody,
suggesting that the peptide is exposed on the virion when incorporated into these regions [48]. Assessed with epitopes of increasing
size, HVR5 was found to accommodate a maximum of 65 amino
acids, while the maximum length accommodated in HVR2 was 33
amino acids [49]. Modifications in HVR1 or 5 reduced susceptibility to neutralization by preexisting immunity (PEI) to the Ad vector
[41,50]. Substituting all the HVR loops in Ad5 with those derived
from Ad43, a serotype with a low prevalence, produced a vector
capable of escaping neutralization with anti-Ad5 sera from mouse,
rabbit and humans [51] and that was still highly immunogenic in
the presence of PEI to the WT virus.
The first capsid display recombinants incorporated 8 amino
acids of the poliovirus type 3 VP1 capsid protein into regions now
recognized as HVR1/2. Antiserum raised against the rAd recognized the poliovirus capsid itself [52]. Worgall et al. incorporated
an immunodominant peptide from the outer membrane protein F
(OprF) of Pseudomonas aeruginosa into HVR5 [43,53]. Immunization
with this rAd induced IgG1 and IgG2a antibody subtypes, elicited
epitope-specific CD4+ and CD8+ T cell responses, and was capable
of protecting 60–80% of mice from a lethal pulmonary challenge
with three different P. aeruginosa strains. Efficacy was increased
with subsequent boosts [43,53]. In contrast, a B-cell epitope from
Bacillus anthracis protective antigen (PA), a subunit of the lethal
toxin, incorporated into HVR5, induced non-neutralizing antibodies and failed to protect against a challenge with lethal toxin [54].
The discordant results from these studies may reflect differential
antibody titers or differing properties of the selected epitopes.
Subsequently, Shiratsuchi et al., inserted a B cell epitope from
the circumsporozoite protein (CSP) of the murine malaria parasite
Plasmodium yoelii into hexon HVR1 or 5 in a recombinant that also
expressed CSP as a transgene [41]. The HVR1 recombinant induced
Co
pi
aa
ut
or
iz
ad
cleared viremia or controlled viremia at the threshold of detection
40 weeks post-challenge.
In these studies, only 35% of macaques exhibiting rAd shedding
[30], suggesting that the protocol used, bicarbonate neutralization of the stomach prior to virus delivery, might not preserve rAd
infectivity. Use of enteric-coated capsules for virus administration
resulted in shedding virus in stool samples of 100% of immunized
macaques [33] emphasizing the importance of an optimal oral
delivery method.
Recently, phase I clinical trial data has been presented for
a transgene-type replication-competent rAd4 vaccine (rAd4-H5Vtn) expressing influenza H5 hemagglutinin (HA) [34]. This virus,
which induced protective immune responses in a nonpermissive
mouse model [35], contains an insertion of the H5 HA gene in place
of part of E3. 166 healthy volunteers received vaccine dosages ranging from 107 to 1011 recombinant virus particles (VP) [34]. Each
cohort received three rAd vaccinations orally and an intramuscular
boost with inactivated H5N1 vaccine. Administration of the rAd was
associated with mild headache, abdominal pain, nasal congestion
and diarrhea. There was no confirmed transmission of the rAd4H5-Vtn virus to household contacts. Pre-existing antibody to Ad4
was associated with a lower immune response to the vaccine, but
this effect was overcome in the high-dose cohorts of 1010 and 1011
VP. In mice, this recombinant elicits good humoral Ad4 and HA
responses but a low cell-mediated response [35]. In humans, the
vaccine induced a significant level of Ad4 seroconversion and HAspecific cellular immune responses in 70% of volunteers receiving
1011 VP [34]. However, HA-specific antibody responses assessed
by hemagglutination-inhibition (HAI) were minimal at all doses
tested, with seroconversion in 4–19% of vaccinated volunteers.
Plasma IgA ELISA titers mirrored HAI, although IgG ELISA responses
indicated 50% seroconversion in the 1011 VP cohort. The H5 HA
antigen is an intrinsically poor immunogen [36], however following
boost of the inactivated H5N1 vaccine, 80–100% of volunteers seroconverted and 80–89% demonstrated antibody titers high enough
to be considered protective in the 1010 and 1011 VP cohorts, respectively [34]. This indicates that although the Ad4-H5-Vtn vaccine
can induce a cellular response, it is only capable of priming an
HA-specific antibody response. The cellular immune response and
replication of the vaccine as assessed by Ad4 seroconversion or PCR
positive rectal swabs, primarily occurred after the first dose, suggesting that only one oral dose may be necessary to induce a cellular
response and prime an antibody response.
The doses of Ad4-H5-Vtn required to induce vector immune
responses are 100-fold (or more) greater than that in the Ad4 vaccine (105 –107 TCID50 [5]). rAd4-H5-Vtn lacks E3, which functions
in evading the host immune response [37] and may contribute to
the vigor or duration of replication, and thus to the immunogenicity
of the, WT vaccines. That possibility has not been experimentally
addressed.
Numerous clinical trials of replicating oncolytic rAds have been
conducted. In general, these studies do not include analyses of
immune responses. Where vector responses have been measured
they are efficiently induced [38,39], but there are no reports of
responses to transgene products.
ap
3238
3. Replicating capsid display rAds as vaccines
Despite the efficacy of the oral Ad4 and Ad7 vaccines and efficient induction of antibodies against the vector, oral rAd vectors
induce only modest antibody responses to transgene products
in both replicating and non-replicating forms [23,40] (Berg and
Ketner, unpublished). However, a second rAd antigen expression
method may offer a more potent approach to induction of humoral
immunity. In capsid-display recombinants, segments of foreign
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
3239
Table 1
Capsid protein insertion sites.
Capsid protein
Insertion sites
Hexon
HVR1
HVR2
HVR3
HVR4
HVR5
HVR6
HVR7
HVR8
HVR9
VR1a
VR4a
Penton
Fiber
RGD loop
C terminus
CD loop
DE loop
FG loop
HI loop
IJ loop
C terminus
pIX
24
45
6
15
66
6
15
–
–
24
24
[41,50–52,55,56,58]
[48,49,51,52,56,58,90,91]
[48,51]
[51,56]
[43,48–51,53,54,56,58,60,63]
[48,51]
[48,51,56]
[51]
[51]
[57]
[57]
9
14
14
14
16
22
16
393
[60]
[64,92–94]
[64]
[64]
[64,95]
[43,60,62–68,95]
[95]
[42,60,71–75,96]
DR
Non-human adenovirus serotype with only 5 variable regions.
Size in amino acids.
or
C
penton base was accessible to anti-HA antibodies, confirming surface location [60]. However, anti-HA antibodies were not detected
in mice immunized with a penton base recombinant containing HA
inserted into the RGD loop [60]. The insertion decreased infectivity
for DC’s, potentially by interfering with integrin binding, which is
involved in virion internalization.
3.3. Fiber (polypeptide IV)
Co
pi
aa
ut
or
iz
ad
high titer antibodies even in mice pre-immunized with WT Ad, suggesting that alteration of HVR1 allowed for evasion of neutralizing
Ad antibodies. An rAd incorporating a B-cell epitope from Plasmodium falciparum CSP in HVR1 induced high-titer antibodies in
mice that recognized parasites expressing the P. falciparum CSP and
neutralized sporozoites bearing the P. falciparum CSP gene in vitro
[55].
The location of epitopes inserted in hexon is an important determinant of immunological properties [49]. rAds that displayed an
epitope from the VP1 capsid protein of Enterovirus 71 in HVRs 1, 2,
or 7 were viable and protected neonatal mice from lethal challenge
through passive immunization and maternally acquired antibodies [56]. However, antibody isotype depended on the location of
the epitope insertion: insertions into HVR1 induced mostly IgG2a
antibodies (Th1) while an HVR7 insertion predominantly produced
IgG1 antibodies (Th2), demonstrating that insertion sites on hexon
are not immunologically equivalent [56]. Similarly, insertion of
the conserved extracellular domain of matrix protein 2 (M2e) of
influenza A virus into variable region 1 (VR1) or VR4 of hexon of
the chimpanzee-origin adenovirus SAd-V25 (AdC68), only the VR1
recombinant provided partial protection from a lethal influenza
challenge [57]. Capsid display recombinants induced more robust
responses than a transgene type recombinant expressing an M2e
fusion protein, supporting the hypothesis that antibody responses
are best induced by antigen displayed in a repetitive and structured
fashion to allow for cross-linkage of the B cell receptors.
Recent studies of rAds with modifications in two hexon HVRs
have demonstrated the potential for single recombinants to elicit
simultaneous antibody responses against two distinct epitopes
[58]. ‘Multivalent’ capsid display recombinants offer potential for
broadening immune responses or inducing responses to genetically
variable pathogens. However, recombinants with different combinations of modified HVRs induced strikingly different responses,
indicating that the design of effective multivalent hexon-modified
rAds may not be straightforward.
References
ap
a
b
Maximum foreign epitope insertion sizeb
3.2. Penton base (polypeptide III)
The penton base and fiber form the penton complex present at
the 12 vertices of the capsid (Fig. 1A). Each penton base monomer
(∼570 residues) contains an Arg-Gly-Asp (RGD) integrin-binding
motif located within a flexible loop at the capsid surface [59].
An influenza A virus HA epitope inserted into the RGD loop of
Fibers are homotrimers of the fiber protein (polypeptide IV) that
protrude from the 12 vertices of the Ad virion and are responsible
for attachment to the host cell (Fig. 1A). The fiber protein has 3
domains: an N-terminal domain that attaches to the penton base, a
central shaft with repeating motifs, and a C-terminal globular knob
responsible for virus attachment to the host cell (Fig. 1E). Ad5 fiber
contains 582 amino acids and is 35–40 nM in length, but fiber length
varies among serotypes due to differing numbers of repeats in the
fiber shaft.
The crystal structure of the fiber knob reveals that the HI loop
(Fig. 1C and E) does not contribute to intramolecular interactions
within the knob, consists mostly of hydrophilic amino acid residues,
is exposed on the surface of the knob and is not involved in the
formation of cell-binding sites [61]. A FLAG epitope inserted into
the HI loop was also accessible to anti-FLAG antibodies, confirming
that the HI loop is exposed [62]. Therefore, the HI loop is seen as
particularly suitable for manipulation and most modifications initially were made at this location [60,63]. More recently, a series of
rAds with insertions of the P. aeruginosa OprF Epi8 epitope in fiber
loops CD, DE, FG, HI and at the C terminus [64] have been examined
for effects of insertions on viral growth in vitro and for immunogenicity. Incorporation of Epi8 into the FG and HI loops had little
effect on viral growth whereas insertion into the CD and DE loops
or at the C terminus strongly reduced infectivity. FG and HI loop
insertions also elicited the strongest humoral and cell-mediated
immune responses and were partially protective against challenge
[64].
Fiber is a target for neutralizing antibodies and substitutions can
contribute to evasion of PEI. For example, modification of the HI
loop circumvented nAb present in ascites fluid from ovarian cancer
patients [65]. Consistent with this, FG and HI loop recombinants
were more effective at inducing antibody and protection in the
presence of PEI than was a transgene-type recombinant expressing all of OprF [64]. The ability to manipulate fiber at multiple sites
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
Co
pi
aa
ut
or
iz
ad
ap
or
C
DR
3240
Fig. 1. Adenovirus capsid structure. (A) Cartoon diagram of an adenovirus particle depicting capsid proteins and DNA. (B) Surface model of the trimeric Ad5 hexon protein
showing the HVR regions. The amino acid location of the region is located in parenthesis. Blue: HVR1 (137–181), red: HVR2 (187–193), yellow: HVR3 (211–218), green: HVR4
(247–260), purple: HVR5 (267–282), brown HVR6 (304–315), orange: HVR7 (418–428), pink: HVR8 (435–436), cyan: HVR9 (440–451). Produced using program PYMOL and
PDB 3IYN [97]. C) Surface model of the trimeric Ad2 penton knob and shaft. Red: FG loop (488–515), blue: HI loop (537–549), green: IJ loop (558–572). Produced using
program PYMOL and PDB 1QIU [98]. D) Map of the Ad5 hexon protein with labeled HVRs E) Map of the fiber protein with labeled ␤-strands in black boxes and FG, HI and IJ
loops indicated with colored arrows.
to allow for the efficacy in the presence of PEI makes fiber insertions
a promising modification for capsid-display vaccines.
Fiber modifications intended to redirect or ablate virus binding
to specific cellular receptors have also been explored [43,66–68].
However, immunogenicity generally is not addressed in those studies.
attach large polypeptides including fluorescent proteins, fully functional enzymes and foreign antigens in viable rAds [71–75]. pIX
fusions containing the envelope protein gp70 of the Friend murine
leukemia virus (FV) [75] and the Yersinia pestis V and F1 capsular antigens [42] induced high-titer antibodies. The ability of pIX
to accommodate very large proteins makes it an attractive site for
display of conformational epitopes.
3.4. pIX (polypeptide IX)
3.5. Comparative immunogenicity
pIX (approximately 140 amino acids) is present in about
240 copies per virion. Trimers of pIX contribute to stability of the
virus particle [69,70]. The C-terminus of pIX is exposed on the surface of the virion and has been used as a substrate on which to
The immunogenicity of influenza A virus HA epitopes inserted
into various Ad capsid proteins has been compared [60]. Insertion
sites included hexon HVR5, the RGD loop of penton base, the HI
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
3241
[41], and such hybrid rAds offer a potential route to greater potency
than either approach alone. Multiple-antigen hybrid rAds, in particular, may be capable of increasing the breadth of immune responses
to pathogens with a high mutation rate, such as influenza A, or a
complex biology, such as malaria. Continued innovation in vaccine
research is critical in order to control diseases such as HIV, influenza
and malaria that have proven refractory to conventional immunization strategies, and replicating rAds have earned their place among
the novel strategies worthy of exploration.
loop of the fiber knob and the C terminus of pIX. All HA insertions
were located on the virion surface, however, an anti-HA antibody
demonstrated strongest binding to HA incorporated into hexon.
Infection of A549 cells and DCs showed that HA incorporation into
hexon interferes minimally with virus entry in vitro, whereas incorporation into fiber knob, pIX and penton base partially reduced
the intracellular Ad genome copy numbers following infection. The
humoral immune response was strongest against the hexon insertion when immunizing with the same number of particles but fiber
was the most immunogenic when controlling for the number of
HA copies per virion [60]. A comparison of an ovalbumin (OVA)
epitope inserted into the fiber HI loop or hexon HVR5 indicated
that fiber insertions were better detected in native virions and triggered a more dramatic increase in anti-OVA antibody responses
upon re-administration [63].
Author contributions
C.D. wrote the paper and performed research on the topic. A.P.
and G.K. contributed to the conception and revising of the article.
Conflict of interest
3.6. Pre-existing immunity and replicating rAds
The authors have no competing financial interests to declare.
DR
Acknowledgements
ap
or
C
This work was funded by NIH grant 1R01AI079132, the Johns
Hopkins Malaria Research Institute, the Marjorie Gilbert Foundation, the Eliasberg Foundation and C.D. was funded through the
Johns Hopkins Sommer Scholars. I would also like to thank Dr. Kasey
Karen and Laura Gelston for their thoughtful insight and helpful
review of this article.
References
Co
pi
aa
ut
or
iz
ad
Antibodies to many Ad serotypes are prevalent in the human
population. PEI to the vaccine serotype can interfere with a
robust immune response against the foreign antigen even in
non-replicating rAds [23,76], although mucosally administered
replication-defective rAd vaccines have elicited transgene-specific
antibodies despite the presence of PEI, and homologous serotype
boosts can be effective [32,77,78]. Importantly, if capable of suppressing the growth of viable rAds, PEI might mitigate the inherent
advantage of vaccine vector replication after administration of a
low dose [79], and live rAds thus may be more sensitive to PEI than
their defective counterparts. PEI can be addressed by use of uncommon human adenovirus serotypes or viruses from other species
[80–82] as vectors. Additionally, as noted above, modifications to
both hexon and fiber have been shown to reduce susceptibility to
PEI [41,50,51] and properly designed capsid display rAds therefore
may be inherently resistant to PEI. Limited experience with replicating rAds has provided some data on the effects of PEI [34], but
this topic must be more thoroughly addressed.
Safety of replicating rAds. Concerns have been expressed over
the safety of replicating vaccines due to the possibility of inducing disease in the immunocompromised and to the possibility of
unintentional spread to contacts. Systemic adenovirus infections
can be fatal in people who are profoundly immunocompromised,
for example, in the course of bone marrow transplantation [83,84].
Further, Ad is commonly present in AIDS-related deaths, although
it is not generally believed that it was the cause [85,86]. Clearly,
live rAds cannot be administered to the severely immunodeficient.
However, unwitting administration of the military vaccine to a
small number of recruits with early HIV infection produced no
observed ill effects, nor did the concurrent HIV infection prolong
shedding, suggesting a small margin of safety in that population
[87]. Transmission of the oral military Ad vaccine did occur, but
required intimate contact, as it was not observed among recruits in
the barracks [88,89], and no confirmed transmission of the rAd4H5-Vtn virus to contacts was observed in its recent clinical trial [34].
This aspect of use of live vaccines, rAd or others, must be carefully
investigated.
4. Conclusion
Replication-competent transgene or capsid display rAds delivered orally to the gut mucosa offer an unconventional immunization approach. Transgene rAds have been shown to induce
a robust cellular-mediated immune response, and capsid-display
rAds promise to induce strong humoral responses. Critically,
transgene and capsid display designs possess complementary
immunological characteristics and can be combined in single rAds
[1] Bourinbaiar AS, Metadilogkul O, Jirathitikal V. Mucosal AIDS vaccines. Viral
Immunol 2003;16(4):427–45.
[2] Silin DS, Lyubomska OV, Jirathitikal V, Bourinbaiar AS. Oral vaccination: where
we are? Expert Opin Drug Deliv 2007;4(4):323–40.
[3] FDA. Complete List of Vaccines Licensed for Immunization and Distribution in the United States; 2013. Available from http://www.fda.gov/
BiologicsBloodVaccines/default.htm [cited 26.04.13].
[4] Lycke N. Recent progress in mucosal vaccine development: potential and limitations. Nat Rev Immunol 2012;12(8):592–605.
[5] Takafuji ET, Gaydos JC, Allen RG, Top Jr FH. Simultaneous administration of live,
enteric-coated adenovirus types 4, 7 and 21 vaccines: safety and immunogenicity. J Infect Dis 1979;140(1):48–53.
[6] Schwartz AR, Togo Y, Hornick RB. Clinical evaluation of live, oral types 1, 2, and
5 adenovirus vaccines. Am Rev Respir Dis 1974;109(2):233–9.
[7] Gutekunst RR, White RJ, Edmondson WP, Chanock RM. Immunization with live
type 4 adenovirus: determination of infectious virus dose and protective effect
of enteric infection. Am J Epidemiol 1967;86(2):341–9.
[8] Top Jr FH, Buescher EL, Bancroft WH, Russell PK. Immunization with live
types 7 and 4 adenovirus vaccines, II. Antibody response and protective
effect against acute respiratory disease due to adenovirus type 7. J Infect Dis
1971;124(2):155–60.
[9] Lyons A, Longfield J, Kuschner R, Straight T, Binn L, Seriwatana J, et al. A doubleblind, placebo-controlled study of the safety and immunogenicity of live, oral
type 4 and type 7 adenovirus vaccines in adults. Vaccine 2008;26(23):2890–8.
[10] Rollier CS, Reyes-Sandoval A, Cottingham MG, Ewer K, Hill AV. Viral
vectors as vaccine platforms: deployment in sight. Curr Opin Immunol
2011;23(3):377–82.
[11] US National Institutes of Health. ClinicalTrials.gov; 2013. Available from
www.clinicaltrials.gov/ [cited 01.05.13].
[12] Lore K, Adams WC, Havenga MJ, Precopio ML, Holterman L, Goudsmit J,
et al. Myeloid and plasmacytoid dendritic cells are susceptible to recombinant
adenovirus vectors and stimulate polyfunctional memory T cell responses. J
Immunol 2007;179(3):1721–9.
[13] Patterson LJ, Kuate S, Daltabuit-Test M, Li Q, Xiao P, McKinnon K, et al. Replicating adenovirus-simian immunodeficiency virus (SIV) vectors efficiently prime
SIV-specific systemic and mucosal immune responses by targeting myeloid
dendritic cells and persisting in rectal macrophages, regardless of immunization route. Clin Vaccine Immunol 2012;19(5):629–37.
[14] Small JC, Ertl HC. Viruses – from pathogens to vaccine carriers. Curr Opin Virol
2011;1(4):241–5.
[15] Huang MM, Hearing P. Adenovirus early region 4 encodes two gene products
with redundant effects in lytic infection. J Virol 1989;63(6):2605–15.
[16] Weinberg DH, Ketner G. Adenoviral early region 4 is required for efficient viral
DNA replication and for late gene expression. J Virol 1986;57(3):833–8.
[17] Jones N, Shenk T. Isolation of deletion and substitution mutants of adenovirus
type 5. Cell 1978;13(1):181–8.
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
or
C
DR
[42] Boyer JL, Sofer-Podesta C, Ang J, Hackett NR, Chluchlolo MJ, Senina S, et al. Protective immunity against a lethal respiratory Yersinia pestis challenge induced
by V antigen or the F1 capsular antigen incorporated into adenovirus capsid.
Hum Gene Ther 2010;21(7):891–901.
[43] Worgall S, Krause A, Qiu J, Joh J, Hackett NR, Crystal RG. Protective immunity to pseudomonas aeruginosa induced with a capsid-modified adenovirus
expressing P. aeruginosa OprF. J Virol 2007;81(24):13801–8.
[44] van Oostrum J, Burnett RM. Molecular composition of the adenovirus type 2
virion. J Virol 1985;56(2):439–48.
[45] Rux JJ, Kuser PR, Burnett RM. Structural and phylogenetic analysis of adenovirus
hexons by use of high-resolution X-ray crystallographic, molecular modeling,
and sequence-based methods. J Virol 2003;77(17):9553–66.
[46] Rux JJ, Burnett RM. Type-specific epitope locations revealed by X-ray crystallographic study of adenovirus type 5 hexon. Mol Ther 2000;1(1):18–30.
[47] Crawford-Miksza L, Schnurr DP. Analysis of 15 adenovirus hexon proteins
reveals the location and structure of seven hypervariable regions containing
serotype-specific residues. J Virol 1996;70(3):1836–44.
[48] Wu H, Han T, Belousova N, Krasnykh V, Kashentseva E, Dmitriev I, et al. Identification of sites in adenovirus hexon for foreign peptide incorporation. J Virol
2005;79(6):3382–90.
[49] Matthews QL, Yang P, Wu Q, Belousova N, Rivera AA, Stoff-Khalili MA, et al.
Optimization of capsid-incorporated antigens for a novel adenovirus vaccine
approach. Virol J 2008;5:98.
[50] Abe S, Okuda K, Ura T, Kondo A, Yoshida A, Yoshizaki S, et al. Adenovirus type
5 with modified hexons induces robust transgene-specific immune responses
in mice with pre-existing immunity against adenovirus type 5. J Gene Med
2009;11(7):570–9.
[51] Bruder JT, Semenova E, Chen P, Limbach K, Patterson NB, Stefaniak ME, et al.
Modification of Ad5 hexon hypervariable regions circumvents pre-existing Ad5
neutralizing antibodies and induces protective immune responses. PLoS ONE
2012;7(4):e33920.
[52] Crompton J, Toogood CI, Wallis N, Hay RT. Expression of a foreign epitope on
the surface of the adenovirus hexon. J Gen Virol 1994;75(Pt 1):133–9.
[53] Worgall S, Krause A, Rivara M, Hee KK, Vintayen EV, Hackett NR, et al. Protection
against P. aeruginosa with an adenovirus vector containing an OprF epitope in
the capsid. J Clin Invest 2005;115(5):1281–9.
[54] McConnell MJ, Danthinne X, Imperiale MJ. Characterization of a permissive
epitope insertion site in adenovirus hexon. J Virol 2006;80(11):5361–70.
[55] Palma C, Overstreet MG, Guedon JM, Hoiczyk E, Ward C, Karen KA, et al.
Adenovirus particles that display the Plasmodium falciparum circumsporozoite
protein NANP repeat induce sporozoite-neutralizing antibodies in mice. Vaccine 2011;29(8):1683–9.
[56] Tian X, Su X, Li X, Li H, Li T, Zhou Z, et al. Protection against enterovirus 71 with
neutralizing epitope incorporation within adenovirus type 3 hexon. PLoS ONE
2012;7(7):pe41381.
[57] Zhou D, Wu TL, Emmer KL, Kurupati R, Tuyishime S, Li Y, et al. Hexon-modified
recombinant E1-deleted adenovirus vectors as dual specificity vaccine carriers
for influenza virus. Mol Ther 2012;21(3):696–706.
[58] Gu L, Li ZC, Krendelchtchikov A, Krendelchtchikova V, Wu H, Matthews
QL. Using multivalent adenoviral vectors for HIV vaccination. PLoS ONE
2013;8(3):e60347.
[59] Zubieta C, Schoehn G, Chroboczek J, Cusack S. The structure of the human
adenovirus 2 penton. Mol Cell 2005;17(1):121–35.
[60] Krause A, Joh JH, Hackett NR, Roelvink PW, Bruder JT, Wickham TJ, et al. Epitopes
expressed in different adenovirus capsid proteins induce different levels of
epitope-specific immunity. J Virol 2006;80(11):5523–30.
[61] Xia D, Henry L, Gerard RD, Deisenhofer J. Structure of the receptor binding domain of adenovirus type 5 fiber protein. Curr Top Microbiol Immunol
1995;199(Pt 1):39–46.
[62] Krasnykh V, Dmitriev I, Mikheeva G, Miller CR, Belousova N, Curiel DT. Characterization of an adenovirus vector containing a heterologous peptide epitope
in the HI loop of the fiber knob. J Virol 1998;72(3):1844–52.
[63] Lanzi A, Ben Youssef G, Perricaudet M, Benihoud K. Anti-adenovirus humoral
responses influence on the efficacy of vaccines based on epitope display on
adenovirus capsid. Vaccine 2010;29(7):1463–71.
[64] Sharma A, Krause A, Xu Y, Sung B, Wu W, Worgall S. Adenovirus-based vaccine
with epitopes incorporated in novel fiber sites to induce protective immunity
against Pseudomonas aeruginosa. PLoS ONE 2013;8(2):e56996.
[65] Blackwell JL, Li H, Gomez-Navarro J, Dmitriev I, Krasnykh V, Richter CA, et al.
Using a tropism-modified adenoviral vector to circumvent inhibitory factors in
ascites fluid. Hum Gene Ther 2000;11(12):1657–69.
[66] Dmitriev I, Krasnykh V, Miller CR, Wang M, Kashentseva E, Mikheeva G,
et al. An adenovirus vector with genetically modified fibers demonstrates
expanded tropism via utilization of a coxsackievirus and adenovirus receptorindependent cell entry mechanism. J Virol 1998;72(12):9706–13.
[67] Wickham TJ, Tzeng E, Shears 2nd LL, Roelvink PW, Li Y, Lee GM, et al. Increased
in vitro and in vivo gene transfer by adenovirus vectors containing chimeric
fiber proteins. J Virol 1997;71(11):8221–9.
[68] Denby L, Work LM, Seggem DJ, Wu E, McVey JH, Nicklin SA, et al. Development of renal-targeted vectors through combined in vivo phage display
and capsid engineering of adenoviral fibers from serotype 19p. Mol Ther
2007;15(9):1647–54.
[69] McConnell MJ, Imperiale MJ. Biology of adenovirus and its use as a vector for
gene therapy. Hum Gene Ther 2004;15(11):1022–33.
[70] Parks RJ. Adenovirus protein IX: a new look at an old protein. Mol Ther
2005;11(1):19–25.
Co
pi
aa
ut
or
iz
ad
[18] Sullivan NJ, Sanchez A, Rollin PE, Yang ZY, Nabel GJ. Development of a preventive vaccine for Ebola virus infection in primates. Nature 2000;408(6812):
605–9.
[19] Thomas MA, Spencer JF, La Regina MC, Dhar D, Tollefson AE, Toth K, et al. Syrian hamster as a permissive immunocompetent animal model for the study of
oncolytic adenovirus vectors. Cancer Res 2006;66(3):1270–6.
[20] Pacini DL, Dubovi EJ, Clyde Jr WA. A new animal model for human respiratory
tract disease due to adenovirus. J Infect Dis 1984;150(1):92–7.
[21] Chengalvala M, Lubeck MD, Davis AR, Mizutani S, Molnar-Kimber K, Morin
J, et al. Evaluation of adenovirus type 4 and type 7 recombinant hepatitis B
vaccines in dogs. Vaccine 1991;9(7):485–90.
[22] Jogler C, Hoffmann D, Theegarten D, Grunwald T, Uberla K, Wildner O, et al.
Replication properties of human adenovirus in vivo and in cultures of primary
cells from different animal species. J Virol 2006;80(7):3549–58.
[23] Lubeck MD, Davis AR, Chengalvala M, Natuk RJ, Morin JE, Molnar-Kimber K,
et al. Immunogenicity and efficacy testing in chimpanzees of an oral hepatitis B vaccine based on live recombinant adenovirus. Proc Natl Acad Sci U S A
1989;86(17):6763–7.
[24] Klessig DF, Grodzicker T. Mutations that allow human Ad2 and Ad5 to express
late genes in monkey cells map in the viral gene encoding the 72K DNA binding
protein. Cell 1979;17(4):957–66.
[25] Toth K, Spencer JF, Tollefson AE, Kuppuswamy M, Doronin K, Lichtenstein DL,
et al. Cotton rat tumor model for the evaluation of oncolytic adenoviruses. Hum
Gene Ther 2005;16(1):139–46.
[26] Tacket CO, Losonsky G, Lubeck MD, Davis AR, Mizutani S, Horwith G, et al. Initial
safety and immunogenicity studies of an oral recombinant adenohepatitis B
vaccine. Vaccine 1992;10(10):673–6.
[27] Kruijer W, van Schaik FM, Sussenbach JS. Structure and organization of the
gene coding for the DNA binding protein of adenovirus type 5. Nucleic Acids
Res 1981;9(18):4439–57.
[28] Buge SL, Richardson E, Alipanah S, Markham P, Cheng S, Kalyan N, et al. An
adenovirus-simian immunodeficiency virus env vaccine elicits humoral, cellular, and mucosal immune responses in rhesus macaques and decreases viral
burden following vaginal challenge. J Virol 1997;71(11):8531–41.
[29] Buge SL, Murty L, Arora K, Kalyanaraman VS, Markham PD, Richardson ES,
et al. Factors associated with slow disease progression in macaques immunized
with an adenovirus-simian immunodeficiency virus (SIV) envelope priminggp120 boosting regimen and challenged vaginally with SIVmac251. J Virol
1999;73(9):7430–40.
[30] Zhao J, Lou Y, Pinczewski J, Malkevitch N, Aldrich K, Kalyanaraman VS, et al.
Boosting of SIV-specific immune responses in rhesus macaques by repeated
administration of Ad5hr-SIVenv/rev and Ad5hr-SIVgag recombinants. Vaccine
2003;21(25–26):4022–35.
[31] Malkevitch N, Patterson LJ, Aldrich K, Richardson E, Alvord WG, Robert-Guroff
M. A replication competent adenovirus 5 host range mutant-simian immunodeficiency virus (SIV) recombinant priming/subunit protein boosting vaccine
regimen induces broad, persistent SIV-specific cellular immunity to dominant and subdominant epitopes in Mamu-A*01 rhesus macaques. J Immunol
2003;170(8):4281–9.
[32] Zhao J, Pinczewski J, Gomez-Roman VR, Venzon D, Kalyanaraman VS, Markham
PD, et al. Improved protection of rhesus macaques against intrarectal simian
immunodeficiency virus SIV(mac251) challenge by a replication-competent
Ad5hr-SIVenv/rev and Ad5hr-SIVgag recombinant priming/gp120 boosting
regimen. J Virol 2003;77(15):8354–65.
[33] Gomez-Roman VR, Grimes Jr GJ, Potti GK, Peng B, Demberg T, Gravlin L, et al.
Oral delivery of replication-competent adenovirus vectors is well tolerated by
SIV- and SHIV-infected rhesus macaques. Vaccine 2006;24(23):5064–72.
[34] Gurwith M, Lock M, Taylor EM, Ishioka G, Alexander J, Mayall T, et al. Safety and
immunogenicity of an oral, replicating adenovirus serotype 4 vector vaccine
for H5N1 influenza: a randomised, double-blind, placebo-controlled, phase 1
study. Lancet Infect Dis 2013;13(3):238–50.
[35] Alexander J, Ward S, Mendy J, Manayani DJ, Farness P, Avanzini JB, et al.
Pre-clinical evaluation of a replication-competent recombinant adenovirus serotype 4 vaccine expressing influenza H5 hemagglutinin. PLoS ONE
2012;7(2):pe31177.
[36] Talaat KR, Karron RA, Liang PH, McMahon BA, Luke CJ, Thumar B, et al. An openlabel phase I trial of a live attenuated H2N2 influenza virus vaccine in healthy
adults. Influenza Other Respi Viruses 2011;7(1):66–73.
[37] Wold WS, Doronin K, Toth K, Kuppuswamy M, Lichtenstein DL, Tollefson AE.
Immune responses to adenoviruses: viral evasion mechanisms and their implications for the clinic. Curr Opin Immunol 1999;11(4):380–6.
[38] Reid T, Galanis E, Abbruzzese J, Sze D, Wein LM, Andrews J, et al. Hepatic
arterial infusion of a replication-selective oncolytic adenovirus (dl1520):
phase II viral, immunologic, and clinical endpoints. Cancer Res 2002;62(21):
6070–9.
[39] Small EJ, Carducci MA, Burke JM, Rodriguez R, Fong L, van Ummersen
L, et al. A phase I trial of intravenous CG7870, a replication-selective,
prostate-specific antigen-targeted oncolytic adenovirus, for the treatment of
hormone-refractory, metastatic prostate cancer. Mol Ther 2006;14(1):107–17.
[40] Lin SW, Cun AS, Harris-McCoy K, Ertl HC. Intramuscular rather than oral administration of replication-defective adenoviral vaccine vector induces specific
CD8+ T cell responses in the gut. Vaccine 2007;25(12):2187–93.
[41] Shiratsuchi T, Rai U, Krause A, Worgall S, Tsuji M. Replacing adenoviral vector HVR1 with a malaria B cell epitope improves immunogenicity
and circumvents preexisting immunity to adenovirus in mice. J Clin Invest
2010;120(10):3688–701.
ap
3242
03/07/2014
C. Deal et al. / Vaccine 31 (2013) 3236–3243
ap
or
C
DR
[84] Baldwin A, Kingham H, Darville M, Foot AB, Grier D, Cornish JM, et al. Outcome
and clinical course of 100 patients with adenovirus infection following bone
marrow transplantation. Bone Marrow Transplant 2000;26(12):1333–8.
[85] Koopmann J, Dombrowski F, Rockstroh JK, Pfeifer U, Sauerbruch T, Spengler U.
Fatal pneumonia in an AIDS patient coinfected with adenovirus and Pneumocystis carinii. Infection 2000;28(5):323–5.
[86] Khoo SH, Bailey AS, de Jong JC, Mandal BK. Adenovirus infections in human
immunodeficiency virus-positive patients: clinical features and molecular epidemiology. J Infect Dis 1995;172(3):629–37.
[87] Rhoads JL, Birx DL, Wright DC, Brundage JF, Brandt BL, Redfield RR, et al. Safety
and immunogenicity of multiple conventional immunizations administered
during early HIV infection. J Acquir Immune Defic Syndr 1991;4(7):724–31.
[88] Mueller RE, Muldoon RL, Jackson GG. Communicability of enteric live adenovirus type 4 vaccine in families. J Infect Dis 1969;119(1):60–6.
[89] Stanley ED, Jackson GG. Spread of enteric live adenovirus type 4 vaccine in
married couples. J Infect Dis 1969;119(1):51–9.
[90] Matthews QL, Fatima A, Tang Y, Perry BA, Tsuruta A, Komarova S, et al. HIV
antigen incorporation within adenovirus hexon hypervariable 2 for a novel
HIV vaccine approach. PLoS ONE 2010;5(7):e11815.
[91] Flatt JW, Fox TL, Makarova N, Blackwell JL, Dmitriev IP, Kashentseva EA, et al.
CryoEM visualization of an adenovirus capsid-incorporated HIV antigen. PLoS
ONE 2012;7(11):e49607.
[92] Wickham TJ, Roelvink PW, Brough DE, Kovesdi I. Adenovirus targeted to
heparan-containing receptors increases its gene delivery efficiency to multiple
cell types. Nat Biotechnol 1996;14(11):1570–3.
[93] Stevenson SC, Rollence M, Marshall-Neff J, McClelland A. Selective targeting
of human cells by a chimeric adenovirus vector containing a modified fiber
protein. J Virol 1997;71(6):4782–90.
[94] Krasnykh VN, Mikheeva GV, Douglas JT, Curiel DT. Generation of recombinant adenovirus vectors with modified fibers for altering viral tropism. J Virol
1996;70(10):6839–46.
[95] Matsui H, Sakurai F, Kurachi S, Tashiro K, Sugio K, Kawabata K, et al. Development of fiber-substituted adenovirus vectors containing foreign peptides in
the adenovirus serotype 35 fiber knob. Gene Ther 2009;16(8):1050–7.
[96] Li J, Le L, Sibley DA, Mathis JM, Curiel DT. Genetic incorporation of HSV-1 thymidine kinase into the adenovirus protein IX for functional display on the virion.
Virology 2005;338(2):247–58.
[97] Liu H, Jin L, Koh SB, Atanasov I, Schein S, Wu L, et al. Atomic structure of human
adenovirus by cryo-EM reveals interactions among protein networks. Science
2010;329(5995):1038–43.
[98] van Raaij MJ, Mitraki A, Lavigne G, Cusack S. A triple beta-spiral in the adenovirus fibre shaft reveals a new structural motif for a fibrous protein. Nature
1999;401(6756):935–8.
Co
pi
aa
ut
or
iz
ad
[71] Li J, Fatima A, Komarova S, Ugai H, Uprety P, Roth JC, et al. Evaluation of adenovirus capsid labeling versus transgene expression. Virol J 2010;7:21.
[72] Meulenbroek RA, Sargent KL, Lunde J, Jasmin BJ, Parks RJ. Use of adenovirus
protein IX (pIX) to display large polypeptides on the virion – generation of fluorescent virus through the incorporation of pIX-GFP. Mol Ther
2004;9(4):617–24.
[73] Kimball KJ, Rivera AA, Zinn KR, Icyuz M, Saini V, Li J, et al. Novel
infectivity-enhanced oncolytic adenovirus with a capsid-incorporated dualimaging moiety for monitoring virotherapy in ovarian cancer. Mol Imaging
2009;8(5):264–77.
[74] Matthews QL, Sibley DA, Wu H, Li J, Stoff-Khalili MA, Waehler R, et al. Genetic
incorporation of a herpes simplex virus type 1 thymidine kinase and firefly
luciferase fusion into the adenovirus protein IX for functional display on the
virion. Mol Imaging 2006;5(4):510–9.
[75] Bayer W, Tenbusch M, Lietz R, Johrden L, Schimmer S, Uberla K, et al. Vaccination with an adenoviral vector that encodes and displays a retroviral antigen
induces improved neutralizing antibody and CD4+ T-cell responses and confers
enhanced protection. J Virol 2010;84(4):1967–76.
[76] Robert-Guroff M. Replicating and non-replicating viral vectors for vaccine
development. Curr Opin Biotechnol 2007;18(6):546–56.
[77] Xiang ZQ, Gao GP, Reyes-Sandoval A, Li Y, Wilson JM, Ertl HC. Oral vaccination
of mice with adenoviral vectors is not impaired by preexisting immunity to the
vaccine carrier. J Virol 2003;77(20):10780–9.
[78] Peters W, Brandi JR, Lindbloom JD, Martinez CJ, Scalian CD, Trager GR, et al.
Oral administration of an adenovirus vector encoding both an avian influenza
A hemagglutinin and a TLR3 ligand induces antigen specific granzyme B and
IFN-gamma T cell responses in humans. Vaccine 2013;31(13):1752–8.
[79] Wohlfart C. Neutralization of adenoviruses: kinetics, stoichiometry, and mechanisms. J Virol 1988;62(7):2321–8.
[80] Xiang Z, Gao G, Reyes-Sandoval A, Cohen CJ, Li Y, Bergelson JM, et al. Novel,
chimpanzee serotype 68-based adenoviral vaccine carrier for induction of antibodies to a transgene product. J Virol 2002;76(6):2667–75.
[81] Dudareva M, Andrews L, Gilbert SC, Bejon P, Marsh K, Mwacharo J, et al. Prevalence of serum neutralizing antibodies against chimpanzee adenovirus 63 and
human adenovirus 5 in Kenyan children, in the context of vaccine vector efficacy. Vaccine 2009;27(27):3501–4.
[82] Kahl CA, Bonnell J, Hiriyanna S, Fultz M, Nyberg-Hoffman C, Chen P, et al. Potent
immune responses and in vitro pro-inflammatory cytokine suppression by a
novel adenovirus vaccine vector based on rare human serotype 28. Vaccine
2010;28(35):5691–702.
[83] Blanke C, Clark C, Broun ER, Tricot G, Cunningham I, Cornetta K, et al. Evolving
pathogens in allogeneic bone marrow transplantation: increased fatal adenoviral infections. Am J Med 1995;99(3):326–8.
3243
03/07/2014